Adhesion molecules and mechanotransduction in vascular physiology and disease

Daniel E. Conway1 and Martin A. Schwartz1,2,3
1
Cardiovascular Research Center and Department of Microbiology
2 Departments of Cell Biology and Biomedical Engineering, University of Virginia, Charlottesville VA 22908
3 Author for correspondence: maschwartz@virginia.edu

Current address: Yale Cardiovascular Research Center, Yale School of Medicine, martin.schwartz@yale.edu

  • Main Image Navi
Endothelial cells respond to hemodynamic mechanical forces such as shear stress and cyclic strain (Fig 1). Disturbances in these forces are associated with endothelial dysfunction and cardiovascular disease. Endothelial cell-cell junctions have been shown to be an important site for transduction of mechanical forces into intracellular signaling events. Several early responses to shear stress occur through a mechanosensory complex, consisting of PECAM-1, VE-cadherin, and VEGFR2, located at the cell-cell junction. This complex is required for the activation of shear-dependent signaling pathways that mediate both cell alignment and activation of many of the pathways that promote atherosclerosis. In this complex, PECAM-1 is hypothesized to be the primary mechanotransducer, where it is directly stimulated by force to initiate signaling. VE-cadherin functions as an adapter that brings VEGFR2 into proximity with PECAM-1, where VEGFR2 is activated by a src family kinase. Activated VEGFR2 then recruits PI3-kinase and mediates activation of downstream pathways. Recent work has revealed additional functions of these proteins in vascular remodeling and function, and shed new light on their involvement in mechanotransduction.
Highlights
  • Locations of atherosclerosis correlated to regions of disturbed flow, which act on the endothelium to stimulate inflammatory pathways
  • A mechanosensory complex at cell junctions consisting of PECAM-1, VE-cadherin, and VEGFR2, plays an important role in sensing flow
  • New evidence supports a role for PECAM-1 in sensation of flow and vascular remodeling in vivo
  • VE-cadherin and VEGFR2 are implicated in force-dependent signaling events in vascular physiology and pathology
Introduction
Signaling in response to flow
Mechanotransducers 
PECAM-1 
VE-cadherin
VEGFR2
Remaining questions
References
Back to top Introduction
Mechanical forces from blood flow directly control the physiology of the cardiovascular system (1). The major forces of physiological and pathological interest are wall shear stress, acting on the endothelium, and circumferential strain, acting on both endothelial and smooth muscle cells (see figure Forces affecting endothelial cells). A major reason for current interest in these forces by both basic scientists and physicians is that sites of atherosclerosis are non-random, strongly correlating with curved or branched sites in the vasculature which, as a result of their geometry, experience low time-averaged shear stress, shear stress reversal, and spatial and temporal gradients in shear stress (2, 3) (see figure Forces affecting endothelial cells). While numerous risk factors for cardiovascular disease are known, including high LDL cholesterol, diabetes and circulating inflammatory mediators, these are relatively uniform throughout the circulation. The local nature of atherosclerosis has therefore led to the concept that biomechanical factors are crucial. High blood pressure also affects local cellular homeostasis and contributes to the development of atherosclerosis and other pathologies. Recent advances in this field have led to the identification of candidate mechanoreceptors, the specific proteins which sense and respond to mechanical forces, and development of viable hypotheses concerning their involvement in human disease. This article will review the basic features of mechanotransduction in the vascular system, focusing on molecular mechanisms and their potential exploitation in clinical research.
Back to top Signaling in response to flow
Endothelial cells, exposed to arterial levels of steady or pulsatile unidirectional shear stress in vitro, typically 10 to 40 dynes/cm2, adopt an anti-inflammatory, anti-thrombotic and anti-proliferative phenotype compared to cells in static culture (4–7). These observations have led to the concept that high shear, typical of straight segments of arteries, protects against atherogenic stimuli. By contrast, low flow, oscillatory flow or other flow patterns that involve changes in flow direction and magnitude induce a pro-inflammatory, pro-thrombotic state characterized by high cell turnover (both proliferation and death) relative to cells in static medium. Thus, flow patterns that mimic those found at atherosclerosis-resistant vs. prone regions of arteries inhibit or promote events involved in development of atherosclerosis, respectively.

Endothelial responses to fluid shear stress are often studied by acute application of flow to naive monolayers of cells. This protocol does not mimic any events in normal physiology but is a convenient and effective way to study mechanisms. The approach is analogous to treating starved cells with a bolus of growth factor to study signaling by growth factor receptors, or replating of suspended cells on extracellular matrix to study integrin signaling. Acute responses to arterial shear stress include release of signaling molecules such as nitric oxide and prostacyclin, activation of GTPases, activation of tyrosine and serine/threonine kinases, and phosphorylation of membrane proteins (8, 9). Released signaling molecules, such as prostacyclin and nitric oxide, are physiologically important as they act on smooth muscle cells to mediate vasorelaxation. However, onset of shear also transiently activates a number of inflammatory pathways, including NF-kB, c-jun N-terminal kinase (JNK), p21-activated kinase (PAK) and reactive oxygen species (ROS).

Endothelial cells exposed to long-term arterial levels of unidirectional shear stress remodel their cytoskeleton and downregulate inflammatory pathways. Remodeling involves cell elongation, alignment of actin stress fibers and focal adhesions in the direction of flow, and movement of the microtubule organizing center toward the downstream side of the nucleus (10, 11). Endothelial cells therefore adapt to unidirectional flow, which appears to be important in the acquisition of a quiescent, atherosclerosis-resistant phenotype.

A second important set of flow responses involves activation of atherosclerosis-protective pathways. Flow activates Erk5, which leads to increased expression of the transcription factor Kruppel-like factor (KLF) 2 (12, 13). This transcription factor induces expression of a large repertoire of atherosclerosis-protective genes, including anti-inflammatory, anti-oxidative and anti-thrombotic proteins. Unlike the inflammatory pathways, this set of signals is sustained in high shear, reaching a maximum only after more than 24 hours.
Back to top Mechanotransducers
Previous studies have proposed that cell-cell junctions (14), heterotrimeric G-proteins (15), primary cilia (16), caveolae (17), integrins (18), and the glycocalyx (19) mediate transmission or transduction of force from fluid shear stress into signals that regulate the endothelium. At present, the primary mechanotransducers responsible for activation of the anti-atherosclerotic pathways such as Erk5 and KLF2 are unknown. However, work over the past decade has identified a mechanosensory complex at cell-cell junctions, consisting of PECAM-1, VE-cadherin, and VEGFR2, which is required for the activation of a number of shear-dependent signaling pathways that mediate both cell alignment and activation of many of the pro-atherosclerotic pathways (14). In this model, PECAM-1 is the primary mechanotransducer that is stimulated by force to initiate signaling. VE-cadherin functions as an adapter that brings VEGFR2 into proximity of PECAM-1, thereby facilitating VEGFR2 transactivation by a src family kinase. Activated VEGFR2 recruits PI3-kinase, and mediates activation of Akt and eNOS (14, 20). Discussion of these pathways will be the main subject of the rest of this review.
Back to top PECAM-1
PECAM-1 is an adhesion and signaling molecule expressed on the surface of endothelial cells, platelets, and leukocytes (21). It is a transmembrane protein with an extracellular region that contains six immunoglobulin-like domains, a transmembrane domain, and a short, highly conserved cytoplasmic tail that contains an immunomodulatory ITIM motif (22) (see figure VE-cadherin and PECAM-1). This motif consists of two tyrosine residues that upon phosphorylation bind the src homology region 2 domain-containing phosphatase 2 (SHP2), which inhibits a number of activating pathways. In endothelial cells, PECAM-1 primarily localizes to intracellular junctions, where it engages in homophilic, trans interactions with PECAM-1 on neighboring cells. It also localizes to the lateral border recycling compartment (23). PECAM-1 has also been found to bind to a number of other counter receptors including integrin αvβ3 but the function of these interactions is less well characterized (24).

PECAM-1 knockout mice have no major developmental or vascular defects (25, 26). However, they have larger diameter collateral arteries compared to wild-type mice (27). A more pronounced decrease in leukocyte emigration was observed in PECAM-1 knockout in FVB/n mice as compared to the standard C57BL/6 strain, suggesting that C57BL/6 mice can compensate for the lack of PECAM-1 (28). Platelets and leukocytes from PECAM-1-/- mice are hyper-activated, most likely due to the loss of the ITIM function (29, 30). Arterioles from PECAM knockout mice exhibit reduced NO-dependent dilation by shear stress (31). In a model of hind limb ischemia, PECAM knockout mice show reduced collateral remodeling and reduced recovery of perfusion, which was attributed to a lack of endothelial response to shear stress (27).

PECAM-1 knockout mice, when crossed into atherosclerotic mouse models, have altered distributions of atherosclerotic lesions. In both the apolipoprotein E (-/-) and the low density lipoprotein receptor (-/-) background, deletion of PECAM decreased lesion size in the lesser curvature of the aortic arch, a principal region of reversing shear stress (32–34). This effect was due to endothelial PECAM-1 since it was also seen in bone marrow chimeras when the donor was PECAM-1-positive. However, PECAM expression was shown to have no effect on lesions in the descending thoracic and abdominal aorta (33). In low density lipoprotein receptor (-/-) mice, PECAM deletion resulted in enlarged lesions in the aortic sinus, branching arteries, and descending thoracic and abdominal aorta (32). Taken together, these studies suggest that PECAM-1 exerts complex effects depending on the model and the site. The data support the idea that endothelial PECAM-1 is an important pro-atherosclerotic mechanosensor for the aortic arch. However, at other sites it can drive both pro- and anti-atherogenic responses. Its immunomodulatory function in platelets and leukocytes is the primary candidate for the anti-atherogenic effects. The data therefore also suggest that the relative importance of these different pathways differ depending on the location within the vasculature, for reasons that are currently unknown.

Human single nucleotide polymorphisms of PECAM are associated with early onset of atherosclerosis and increased risk of cardiovascular disease. The SNPs fall into two distinct haplotypes L98S536R643 and V98N536G643, the latter polymorphism being more often associated with cardiovascular disease (35). It is unclear the relative importance of each polymorphic site; however, the R643G dimorphism is in the cytoplasmic domain near the first ITIM site, suggesting that this dimorphism could alter PECAM signaling. Interestingly, endothelial PECAM tyrosine phosphorylation was increased in the homozygous VNG as compared to the homozygous LSR (36, 37). The PECAM polymorphisms have also been shown to affect leukocyte binding to and transmigration through ECs (37, 38). It remains unclear if the PECAM polymorphisms result in increased cardiovascular disease through altered endothelial cell mechanoresponsiveness or increased monocyte/macrophage/platelet reactivity.

The cytoplasmic binding partners of PECAM-1 are at present incompletely characterized. PECAM-1 has been shown to interact with β-catenin and γ-catenin (also known as plakoglobin) (39, 40). However, this has not been studied extensively. The ITIM tyrosines (Y663 and Y686) can be phosphorylated by src family kinases and then binds the protein tyrosine phosphatase SHP2. Fyn appears to be the major kinase responsible for ITIM phosphorylation in response to mechanical stimulation (41). Both shear stress and cyclic stretch result in increased tyrosine phosphorylation of PECAM (41, 42). PECAM phosphorylation and its interaction with SHP2 has been implicated in activation of Erk MAP kinase (43), transactivation of VEGFR2 (44) and downstream events in response to flow (42), as well as modulation of platelet and leukocyte reactivity (30, 45, 46). Phosporylation of PECAM ITIM tyrosines is also implicated in shear stress activation of eNOS, an important anti-inflammatory pathway (42, 44). However, this result is complex since deletion of PECAM increases basal eNOS activity and NO production as well as decreasing stimulation of NO production by flow (47).

Several model systems have been used by the Fujiwara group to study mechanotransduction by PECAM. Stretching confluent monolayers of endothelial cells or exposure to hypoosmotic medium that induces cell swelling also increased PECAM-1 tyrosine phosphorylation (43). Interestingly, PECAM was still phosphorylated by cyclic stretch in detergent extracted cells, indicating that PECAM phosphorylation is not dependent on an intact plasma membrane or soluble cytoplasmic components (41). Pulling on PECAM directly with magnetic beads coated with PECAM antibodies also induced tyrosine phosphorylation, similarly to shear stress (43). However, unlike the response to flow where PECAM is essential for actin alignment parallel to the direction of shear stress (14), PECAM knockout endothelial cells align normally in cyclic stretch (perpendicular to the direction of stretch), demonstrating that PECAM-1 is not required for these responses (27). These data were interpreted to mean that multiple means of mechanically stimulating endothelial cells leads to increased force across PECAM-1 and similar downstream signals.
Back to top VE-cadherin
Vascular endothelial (VE) cadherin is a critical component of endothelial adherens junctions and a determinant of vascular permeability in vitro and in vivo (48). The extracellular domain of VE-cadherin engages in homophilic binding both within one cell (cis interaction) and with neighboring cells (trans interaction). As in other cadherins, this domain is stabilized by direct binding of calcium ions. VE-cadherin has a transmembrane domain followed by a cytoplasmic tail that directly associates with β-catenin and γ-catenin (plakoglobin) (see figure VE-cadherin and PECAM-1). These proteins ultimately link VE-cadherin to the actin cytoskeleton. Classically, it was thought that α-catenin links β-catenin to actin; however, it has recently been suggested that α-catenin cannot simultaneously bind β-catenin and actin, thus, the cytoskeletal linkage must require other components or mechanisms (49). p120-catenin, also binds to the cytoplasmic domain of VE-cadherin and is thought to regulate cadherin function, stability, and availability at the surface (48, 50). VE-cadherin is necessary for proper vascular development, as its loss or expression of a truncated VE-cadherin lacking the beta-catenin binding site are both embryonic lethal, exhibiting severe vascular abnormalities (51).

Application of mechanical force to cadherins stimulates cellular signaling and actin assembly (52, 53), suggesting that cadherins can act as mechanosensors. Adherens junctions have also been shown to strengthen in response to applied mechanical load (54-56). The specific mechanism for this force-dependent strengthening is not fully undestood, although vasodilator-stimulated phosphoprotein (54), α-catenin (57), vinculin (55), myosin (56), and Rac1 (56) have all been shown to participate.

VE-cadherin is implicated in several aspects of the endothelial responses to fluid shear stress. As mentioned above, it is a component of the junctional complex along with PECAM-1 and VE-cadherin. As such, it is necessary for the transactivation of VEGFR2 and association of the adapter Shc with activated VEGFR2 (58). However, homophilic binding of VE-cadherin is not required for its role in response to shear stress (14). These and other data suggest that VE-cadherin functions mainly as an adapter in this system, bringing PECAM-1 and VEGFR2 together to promote activation of the tyrosine kinase following application of force to PECAM-1. This notion is supported by other studies indicating an association and functional relationship between VE-cadherin and VEGFR2 (51).

VE-cadherin also appears to be a downstream target of shear pathways. Endothelial cells exposed in vitro to onset of flow show a transient disruption of cell-cell junctions coincident with increased permeability of tracers across the cell monolayer, followed by recovery at later times (59). Destabilization was mediated by activation of p21-activated kinase, which stimulated myosin-dependent contraction (59) and also directly phosphorylates VE-cadherin to induce its internalization (60). By contrast, cells exposed to reversing (oscillatory) shear stress show sustained activation of PAK and destabilization of junctions (59). This result correlates with the intermittent VE-cadherin staining at the cell-cell junctions and higher permeability in vivo at regions of disturbed shear (61). These results suggest that shear might alter its own mechanotransducers to modulate cellular responses.

Finally, VE-cadherin has also been implicated in responses to mechanical strain. In this system, stretching ECs on an elastic membrane stimulated cell cycle progression which was dependent on homophilic VE-cadherin adhesion (62). This VE-cadherin-dependent effect of stretch was mediated by activation of the small GTPase Rac1. Whether VE-cadherin is the true force bearing element and mechanotransducer in this system was not established; its involvement as a modifier of the mechanotransduction response is also possible, as in the response to flow. A possible role of VE-cadherin in responses of arteries to blood pressure-induced stretch, however, is still very interesting.
Back to top VEGFR2
VEGFR2, also known as KDR (in humans) or FLK-1 (in mice), is the transmembrane receptor tyrosine kinase (RTK) that mediates most of the angiogenic effects of VEGF. VEGFR2 consists of an extracellular ligand-binding domain with seven immunoglobulin (Ig)-like motifs, a single transmembrane domain and a cytoplasmic domain that contains a juxtamembrane region, a kinase domain split by a kinase insert, and a carboxyl terminus. Similar to other RTKs, ligand binding induces dimerization of VEGFR2, transphosphorylation of the receptors, and the association of adapter/signaling proteins to the phosphorylation sites. Knockout of VEGFR2 leads to embryonic death at mouse E 8.5-9.5 due to defective endothelial cell differentiation (63).

A number of studies have implicated VEGFR2 in responses of endothelial cells to shear. Shear stimulates ligand-independent phosphorylation of VEGFR2 and activation of downstream pathways such as Akt and MAP kinases (20, 64, 65). Chemical inhibitors reduced the activation of these pathways, production of nitric oxide, and the phosphorylation and translocation to EC junctions of Shc in response to shear stress (58). Tyrosine phosphorylation of Y801 and Y1175 of the VEGFR2 cytoplasmic tail is essential for this shear stress response, mediating its association with PI3-kinase (14). A chemical inhibitor of VEGFR2 also blocked a transient increase in actin stiffness in cells exposed to shear (66). These results may be explained by the participation of VEGFR2 in the junctional complex with PECAM-1 and VE-cadherin, though other roles cannot currently be excluded.

Caveolin has been implicated in these VEGFR2 pathways as well. Knockdown of caveolin-1 attenuated AKT and ERK1/2 activation by both shear stress and VEGF stimulation (67). Indeed, caveolin-1 has been proposed to be a transducer of mechanical signals in endothelial cells (68, 69). However, loss of caveolin-1 results in drastic de-regulation of eNOS and production of NO, among other pathways (70). Thus, it is difficult to determine to what extent results from caveolin-1 knockout or knockdown are due to changes in mechanotransduction vs. changes in endothelial signaling and gene expression that interfere with the responses measured.

Back to top Remaining questions
Although PECAM-1, VE-cadherin, and VEGFR2 form a mechanosensory complex, almost nothing is known about how these proteins associate, or if there are other proteins associated with the mechanosensory complex. It is notable that N-cadherin, which is also expressed in endothelial cells, does not appear to be able to substitute for VE-cadherin. Thus, VE-cadherin appears to have specific functions not shared with other cadherins. These VE-cadherin specific functions are likely to involve (direct or indirect) interactions with PECAM-1 and VEGFR2. Elucidation of these functions and interactions is a major area for future research.

VEGF-A stimulation was shown to signal through the formation of a VE-cadherin/ β-catenin/PI3-kinase/VEGFR2 complex, similar to the mechanosensory complex (51). There may therefore be a parallel between conventional VEGFR2 signaling and the mechanotransduction pathway, since PI 3-kinase is crucial in both systems. A recent report showed that insulin growth factor-I receptor, E-cadherin, and αv integrin formed a complex with α-catenin, which served as a scaffold (71). These data suggest that synergies between cadherins and growth factor receptors may exist in other systems. Interestingly, a truncated form of VE-cadherin lacking the β-catenin binding region blocked intracellular transmission of the VEGF-A stimulus (51). This result is similar to previous observations that cells lacking β-catenin do not respond to flow (14) and that β-catenin is associated with the mechanosensory complex (14, 65), suggesting that β-catenin is also crucial in both systems.

Another area that needs further exploration is the function of the PECAM cytoplasmic tail. Despite the overwhelming evidence that PECAM is mechanoresponsive, it is difficult to postulate a mechanism for the mechanosensitivity of PECAM if there is no connection to some element of the cytoskeleton. The structure, interactions and mechanosensitity of the PECAM-1 cytosplasmic domain is therefore also an important area.

Finally, it will be important to sort out the multiple functions of PECAM-1 in different cell types in the context of mechanotransduction and atherosclerosis. Tissue-specific, conditional knockout of PECAM-1 in endothelial cells, leukocytes and platelets is needed to resolve the complex results from whole animal knockout studies. Specific mutations that disable individual PECAM functions in mechanotransduction and conventional signaling would also be very useful to address these questions.
Back to top References
  1. Davies, P. F., (2009) Hemodynamic shear stress and the endothelium in cardiovascular pathophysiology. Nat. Clin. Pract. Cardiovasc. Med. 6, 16-26.
  2. Davies, P. F., (2008) Endothelial transcriptome profiles in vivo in complex arterial flow fields. Ann. Biomed. Eng. 36, 563-570.
  3. Glagov, S., C. Zarins, D. P. Giddens, and D. N. 10 Ku, (1988) Hemodynamics and atherosclerosis. Insights and perspectives gained from studies of human arteries. Arch. Pathol. Lab. Med. 112, 1018-31
  4. Chien, S., (2008) Effects of disturbed flow on endothelial cells. Ann Biomed Eng 36, 554-562.
  5. Yee, A., K. A. Bosworth, D. E. Conway, S. G. Eskin, and L. V. McIntire, (2008) Gene expression of endothelial cells under pulsatile non-reversing vs. steady shear stress; comparison of nitric oxide production. Ann. Biomed. Eng. 36, 571-9.
  6. McCormick, S. M., et al., (2001) DNA microarray reveals changes in gene expression of shear stressed human umbilical vein endothelial cells. Proc. Natl. Acad. Sci. USA 98, 8955-60.
  7. Dai, G., et al., (2004) Distinct endothelial phenotypes evoked by arterial waveforms derived from atherosclerosis-susceptible and -resistant regions of human vasculature. Proc. Natl. Acad. Sci. USA 101, 14871-6
  8. White, C. R., and J. A. Frangos, (2007) The shear stress of it all: the cell membrane and mechanochemical transduction. Philos. Trans. R Soc. Lond. B Biol. Sci. 362, 1459-67.
  9. Frangos, J.A., S.G. Eskin, L.V. McIntire, and C.L. 4693 Ives, (1985) Flow effects on prostacyclin production by cultured human endothelial cells. Science 227, 1477-9
  10. Orr, A. W., B. P. Helmke, B. R. Blackman, and M. A. Schwartz, (2006) Mechanisms of mechanotransduction. Dev. Cell. 10, 11-20.
  11. Eskin, S.G. , and L.V. McIntire, (1988) Hemodynamic effects on atherosclerosis and thrombosis. Semin. Thromb. Hemost. 14, 170-4
  12. Fledderus, J. O., et al., (2007) Prolonged shear stress and KLF2 suppress constitutive proinflammatory transcription through inhibition of ATF2. Blood 109, 4249-57.
  13. Parmar, K. M., et al., (2006) Integration of flow-dependent endothelial phenotypes by Kruppel-like factor 2. J. Clin. Invest. 116, 49-58.
  14. Tzima, E., et al., (2005) A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nature 437, 426-31.
  15. Gudi, S., et al., (2003) Rapid activation of Ras by fluid flow is mediated by Galpha(q) and Gbetagamma subunits of heterotrimeric G proteins in human endothelial cells. Arterioscler. Thromb. Vasc. Biol. 23, 994-1000.
  16. Hierck, B. P., et al., (2008) Primary cilia sensitize endothelial cells for fluid shear stress. Dev. Dyn. 237, 725-35.
  17. Yu, J., et al., (2006) Direct evidence for the role of caveolin-1 and caveolae in mechanotransduction and remodeling of blood vessels. J. Clin. Invest. 116, 1284-91.
  18. Jalali, S., et al., (2001) Integrin-mediated mechanotransduction requires its dynamic interaction with specific extracellular matrix (ECM) ligands. Proc. Natl. Acad. Sci. USA 98, 1042-6.
  19. Pahakis, M. Y., J. R. Kosky, R. O. Dull, and J. M. Tarbell, (2007) The role of endothelial glycocalyx components in mechanotransduction of fluid shear stress. Biochem. Biophys. Res. Commun. 355, 228-33.
  20. Jin, Z. G., H. Ueba, T. Tanimoto, A. O. Lungu, M. D. Frame, and B. C. Berk, (2003) Ligand-independent activation of vascular endothelial growth factor receptor 2 by fluid shear stress regulates activation of endothelial nitric oxide synthase. Circ. Res 93, 354-63.
  21. Woodfin, A., M. B. Voisin, and S. Nourshargh, (2007) PECAM-1: a multi-functional molecule in inflammation and vascular biology. Arterioscler. Thromb. Vasc. Biol. 27, 2514-23.
  22. Fujiwara, K., (2006) Platelet endothelial cell adhesion molecule-1 and mechanotransduction in vascular endothelial cells. J. Intern. Med. 259, 373-80.
  23. Mamdouh, Z., X. Chen, L. M. Pierini, F. R. Maxfield, and W. A. Muller, (2003) Targeted recycling of PECAM from endothelial surface-connected compartments during diapedesis. Nature 421, 748-53.
  24. Wong, C. W., et al., (2000) PECAM-1/CD31 trans-homophilic binding at the intercellular junctions is independent of its cytoplasmic domain; evidence for heterophilic interaction with integrin alphavbeta3 in Cis. Mol. Biol. Cell. 11, 3109-21.
  25. Duncan, G. S., et al., (1999) Genetic evidence for functional redundancy of Platelet/Endothelial cell adhesion molecule-1 (PECAM-1): CD31-deficient mice reveal PECAM-1-dependent and PECAM-1-independent functions. J. Immunol. 162, 3022-30.
  26. Mahooti, S., et al., (2000) PECAM-1 (CD31) expression modulates bleeding time in vivo. Am. J. Pathol. 157, 75-81.
  27. Chen, Z., J. Rubin, and E. Tzima, (2010) Role of PECAM-1 in arteriogenesis and specification of preexisting collaterals. Circ. Res. 107, 1355-63.
  28. Schenkel, A. R., T. W. Chew, and W. A. Muller, (2004) Platelet endothelial cell adhesion molecule deficiency or blockade significantly reduces leukocyte emigration in a majority of mouse strains. J. Immunol. 173, 6403-8.
  29. Patil, S., D. K. Newman, and P. J. Newman, (2001) Platelet endothelial cell adhesion molecule-1 serves as an inhibitory receptor that modulates platelet responses to collagen. Blood 97, 1727-32.
  30. Wilkinson, R., A. B. Lyons, D. Roberts, M. X. Wong, P. A. Bartley, and D. E. Jackson, (2002) Platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31) acts as a regulator of B-cell development, B-cell antigen receptor (BCR)-mediated activation, and autoimmune disease. Blood 100, 184-93.
  31. Bagi, Z., J. A. Frangos, J. C. Yeh, C. R. White, G. Kaley, and A. Koller, (2005) PECAM-1 mediates NO-dependent dilation of arterioles to high temporal gradients of shear stress. Arterioscler. Thromb. Vasc. Biol. 25, 1590-5.
  32. Goel, R., et al., (2008) Site-specific effects of PECAM-1 on atherosclerosis in LDL receptor-deficient mice. Arterioscler. Thromb. Vasc. Biol. 28, 1996-2002.
  33. Harry, B. L., et al., (2008) Endothelial cell PECAM-1 promotes atherosclerotic lesions in areas of disturbed flow in ApoE-deficient mice. Arterioscler. Thromb. Vasc. Biol. 28, 2003-8.
  34. Stevens, H. Y., B. Melchior, K. S. Bell, S. Yun, J. C. Yeh, and J. A. Frangos, (2008) PECAM-1 is a critical mediator of atherosclerosis. Dis. Model. Mech. 1, 175-81; discussion 179.
  35. Novinska, M. S., B. C. Pietz, T. M. Ellis, D. K. Newman, and P. J. Newman, (2006) The alleles of PECAM-1. Gene 376, 95-101.
  36. Elrayess, M. A., et al., (2004) R643G polymorphism in PECAM-1 influences transendothelial migration of monocytes and is associated with progression of CHD and CHD events. Atherosclerosis 177, 127-35.
  37. Bayat, B., S. Werth, U. J. Sachs, D. K. Newman, P. J. Newman, and S. Santoso, (2011) Neutrophil transmigration mediated by the neutrophil-specific antigen CD177 is influenced by the endothelial S536N dimorphism of platelet endothelial cell adhesion molecule-1. J. Immunol. 184, 3889-96.
  38. Goodman, R. S., et al., (2008) PECAM-1 polymorphism affects monocyte adhesion to endothelial cells. Transplantation 85, 471-7.
  39. Biswas, P., et al., (2005) Identification of the regions of PECAM-1 involved in beta- and gamma-catenin associations. Biochem. Biophys. Res. Commun. 329, 1225-33.
  40. Biswas, P., et al., (2006) PECAM-1 affects GSK-3beta-mediated beta-catenin phosphorylation and degradation. Am. J. Pathol. 169, 314-24.
  41. Chiu, Y. J., E. McBeath, and K. Fujiwara, (2008) Mechanotransduction in an extracted cell model: Fyn drives stretch- and flow-elicited PECAM-1 phosphorylation. J. Cell. Biol. 182, 753-63.
  42. Fleming, I., B. Fisslthaler, M. Dixit, and R. Busse, (2005) Role of PECAM-1 in the shear-stress-induced activation of Akt and the endothelial nitric oxide synthase (eNOS) in endothelial cells. J. Cell. Sci. 118, 4103-11.
  43. Osawa, M., M. Masuda, K. Kusano, and K. Fujiwara, (2002) Evidence for a role of platelet endothelial cell adhesion molecule-1 in endothelial cell mechanosignal transduction: is it a mechanoresponsive molecule? J. Cell. Biol. 158, 773-85.
  44. Tai, L. K., Q. Zheng, S. Pan, Z. G. Jin, and B. C. Berk, (2005) Flow activates ERK1/2 and endothelial nitric oxide synthase via a pathway involving PECAM1, SHP2, and Tie2. J. Biol. Chem. 280, 29620-4.
  45. Jackson, D. E., C. M. Ward, R. Wang, and P. J. Newman, (1997) The protein-tyrosine phosphatase SHP-2 binds platelet/endothelial cell adhesion molecule-1 (PECAM-1) and forms a distinct signaling complex during platelet aggregation. Evidence for a mechanistic link between PECAM-1- and integrin-mediated cellular signaling. J. Biol. Chem. 272, 6986-93.
  46. Newman, D. K., C. Hamilton, and P. J. Newman, (2001) Inhibition of antigen-receptor signaling by Platelet Endothelial Cell Adhesion Molecule-1 (CD31) requires functional ITIMs, SHP-2, and p56(lck). Blood 97, 2351-7.
  47. McCormick, M. E., R. Goel, D. Fulton, S. Oess, D. Newman, and E. Tzima, (2011) Platelet-endothelial cell adhesion molecule-1 regulates endothelial NO synthase activity and localization through signal transducers and activators of transcription 3-dependent NOSTRIN expression. Arterioscler. Thromb. Vasc. Biol. 31, 643-9.
  48. Vincent, P. A., K. Xiao, K. M. Buckley, and A. P. Kowalczyk, (2004) VE-cadherin: adhesion at arm's length. Am. J. Physiol. Cell. Physiol. 286, C987-97.
  49. Weis, W. I., and W. J. Nelson, (2006) Re-solving the cadherin-catenin-actin conundrum. J. Biol. Chem. 281, 35593-7.
  50. Harris, E. S., and W. J. Nelson, (2010) VE-cadherin: at the front, center, and sides of endothelial cell organization and function. Curr. Opin. Cell. Biol. 22, 651-8.
  51. Carmeliet, P., et al., (1999) Targeted deficiency or cytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediated endothelial survival and angiogenesis. Cell 98, 147-57.
  52. Bard, L., C. Boscher, M. Lambert, R. M. Mege, D. Choquet, and O. Thoumine, (2008) A molecular clutch between the actin flow and N-cadherin adhesions drives growth cone migration. J. Neurosci. 28, 5879-90.
  53. Ko, K. S., P. D. Arora, and C. A. McCulloch, (2001) Cadherins mediate intercellular mechanical signaling in fibroblasts by activation of stretch-sensitive calcium-permeable channels. J. Biol. Chem. 276, 35967-77.
  54. Kris, A. S., R. D. Kamm, and A. L. Sieminski, (2008) VASP involvement in force-mediated adherens junction strengthening. Biochem. Biophys. Res. Commun. 375, 134-8.
  55. le Duc, Q., et al., (2010) Vinculin potentiates E-cadherin mechanosensing and is recruited to actin-anchored sites within adherens junctions in a myosin II-dependent manner. J Cell Biol 189, 1107-15.
  56. Liu, Z., et al., (2010) Mechanical tugging force regulates the size of cell-cell junctions. Proc. Natl. Acad. Sci. USA 107, 9944-9.
  57. Yonemura, S., Y. Wada, T. Watanabe, A. Nagafuchi, and M. Shibata, (2010) alpha-Catenin as a tension transducer that induces adherens junction development. Nat. Cell. Biol. 12, 533-42.
  58. Liu, Y., D. T. Sweet, M. Irani-Tehrani, N. Maeda, and E. Tzima, (2008) Shc coordinates signals from intercellular junctions and integrins to regulate flow-induced inflammation. J Cell Biol 182, 185-96.
  59. Orr, A. W., et al., (2007) Matrix-specific p21-activated kinase activation regulates vascular permeability in atherogenesis. J. Cell. Biol. 176, 719-27.
  60. Gavard, J., and J. S. Gutkind, (2006) VEGF controls endothelial-cell permeability by promoting the beta-arrestin-dependent endocytosis of VE-cadherin. Nat. Cell. Biol. 8, 1223-34.
  61. Miao, H., et al., (2005) Effects of flow patterns on the localization and expression of VE-cadherin at vascular endothelial cell junctions: in vivo and in vitro investigations. J. Vasc. Res. 42, 77-89.
  62. Liu, W. F., C. M. Nelson, J. L. Tan, and C. S. Chen, (2007) Cadherins, RhoA, and Rac1 are differentially required for stretch-mediated proliferation in endothelial versus smooth muscle cells. Circ. Res. 101, e44-52.
  63. Shalaby, F., et al., (1995) Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 376, 62-6.
  64. Chen, K. D., et al., (1999) Mechanotransduction in response to shear stress. Roles of receptor tyrosine kinases, integrins, and Shc. J. Biol. Chem. 274, 18393-400.
  65. Shay-Salit, A., et al., (2002) VEGF receptor 2 and the adherens junction as a mechanical transducer in vascular endothelial cells. Proc. Natl. Acad. Sci. USA 99, 9462-7.
  66. van der Meer, A. D., Y. Li, M. H. Duits, A. A. Poot, J. Feijen, and I. Vermes, (2010) Shear stress induces a transient and VEGFR-2-dependent decrease in the motion of injected particles in endothelial cells. Biorheology 47, 179-92.
  67. van der Meer, A. D., M. M. Kamphuis, A. A. Poot, J. Feijen, and I. Vermes, (2009) Lowering caveolin-1 expression in human vascular endothelial cells inhibits signal transduction in response to shear stress. Int. J. Cell. Biol. 2009, 532432.
  68. Albinsson, S., I. Nordstrom, K. Sward, and P. Hellstrand, (2008) Differential dependence of stretch and shear stress signaling on caveolin-1 in the vascular wall. Am. J. Physiol. Cell. Physiol. 294, C271-9.
  69. Radel, C., M. Carlile-Klusacek, and V. Rizzo, (2007) Participation of caveolae in beta1 integrin-mediated mechanotransduction. Biochem. Biophys. Res. Commun. 358, 626-31.
  70. Balligand, J. L., O. Feron, and C. Dessy, (2009) eNOS activation by physical forces: from short-term regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol. Rev. 89, 481-534.
  71. Canonici, A., et al., (2008) Insulin-like growth factor-I receptor, E-cadherin and alpha v integrin form a dynamic complex under the control of alpha-catenin. Int. J. Cancer. 122, 572-82.


Images
Forces affecting endothelial cells
Forces affecting endothelial cells.
Endothelial cells are exposed to two major forces: cyclic strain [A] and fluid shear stress [B]. [C] ECs in areas of high curvature or downstream of bifurcations (e.g. carotid sinus, coronary bifurcation) are exposed to disturbed shear stress (oscillatory with a low time-average shear stress). (Image created by Ken Mattiuz)
VE-cadherin and PECAM-1
VE-cadherin and PECAM-1.
General structure of VE-cadherin [A] and PECAM-1 [B]. Both transmembrane proteins contain an extracellular region responsible for forming trans homodimers with neighboring cells. The intracellular cytoplasmic tails of each molecule can interact with the actin cytoskeleton. (Image created by Ken Mattiuz)